You are on page 1of 15

Downloaded from http://rsob.royalsocietypublishing.

org/ on May 27, 2017

Over-expression of Plk4 induces


centrosome amplification, loss of primary
rsob.royalsocietypublishing.org
cilia and associated tissue hyperplasia
in the mouse
Paula A. Coelho1, Leah Bury1, Marta N. Shahbazi2, Kifayathullah Liakath-Ali1,3,
Research Peri H. Tate4, Sam Wormald4, Christopher J. Hindley5, Meritxell Huch5,
Cite this article: Coelho PA et al. 2015 Joy Archer6, William C. Skarnes4, Magdalena Zernicka-Goetz2
Over-expression of Plk4 induces centrosome and David M. Glover1
amplification, loss of primary cilia and
1
Department of Genetics, University of Cambridge, Downing Street, Cambridge CB2 3EH, UK
associated tissue hyperplasia in the mouse. 2
Department of Physiology, Development and Neuroscience, Physiological Laboratory, University of Cambridge,
Open Biol. 5: 150209. Downing Street, Cambridge CB2 3EG, UK
3
http://dx.doi.org/10.1098/rsob.150209 Centre for Stem Cells and Regenerative Medicine, Kings College London, Floor 28, Tower Wing, Guys Hospital,
Great Maze Pond, London SE1 9RT, UK
4
Wellcome Trust Genome Campus, the Wellcome Trust Sanger Institute, Cambridge, Hinxton CB10 1SA, UK
5
Henry Wellcome Building of Cancer and Developmental Biology, the Wellcome Trust/Cancer Research UK
Gurdon Institute, Tennis Court Road, Cambridge CB2 1QN, UK
Received: 22 October 2015 6
Department of Veterinary Medicine, University of Cambridge, Madingley Road, Cambridge CB3 0ES, UK
Accepted: 2 December 2015
To address the long-known relationship between supernumerary centro-
somes and cancer, we have generated a transgenic mouse that permits
inducible expression of the master regulator of centriole duplication, Polo-
Subject Area: like-kinase-4 (Plk4). Over-expression of Plk4 from this transgene advances
the onset of tumour formation that occurs in the absence of the tumour sup-
developmental biology/cellular biology
pressor p53. Plk4 over-expression also leads to hyperproliferation of cells in
the pancreas and skin that is enhanced in a p53 null background. Pancreatic
Keywords: islets become enlarged following Plk4 over-expression as a result of equal
primary cilia, Polo-like-kinase-4, skin, tumour expansion of a- and b-cells, which exhibit centrosome amplification. Mice
development, centrosome amplification, overexpressing Plk4 develop grey hair due to a loss of differentiated melano-
pancreas cytes and bald patches of skin associated with a thickening of the epidermis.
This reflects an increase in proliferating cells expressing keratin 5 in the basal
epidermal layer and the expansion of these cells into suprabasal layers. Such
cells also express keratin 6, a marker for hyperplasia. This is paralleled by a
Author for correspondence: decreased expression of later differentiation markers, involucrin, filaggrin
David M. Glover and loricrin. Proliferating cells showed an increase in centrosome number
e-mail: dmg25@hermes.cam.ac.uk and a loss of primary cilia, events that were mirrored in primary cultures
of keratinocytes established from these animals. We discuss how repeated
duplication of centrioles appears to prevent the formation of basal bodies
leading to loss of primary cilia, disruption of signalling and thereby aberrant
differentiation of cells within the epidermis. The absence of p53 permits cells
with increased centrosomes to continue dividing, thus setting up a neoplastic
state of error prone mitoses, a prerequisite for cancer development.

1. Introduction
The centrosome was first described almost simultaneously by Edouard van
Beneden working in Liege and Theodor Boveri in Munich in 1887 (reviewed
in [1]) as a structure at the poles of the mitotic spindle that persisted through
the life cycle of the cell. Towards the end of the nineteenth century tumour
cells were already observed to have multiple spindle poles, and now we
know supernumerary centrosomes to be present in a wide range of solid and

& 2015 The Authors. Published by the Royal Society under the terms of the Creative Commons Attribution
License http://creativecommons.org/licenses/by/4.0/, which permits unrestricted use, provided the original
author and source are credited.
Downloaded from http://rsob.royalsocietypublishing.org/ on May 27, 2017

haematological tumours, including pancreatic, ovarian, colon Correct centrosome behaviour is also required for the devel- 2
and prostate cancer, multiple myeloma, non-Hodgkins and opment of cerebral cortex of the mammalian brain. Deficiency

rsob.royalsocietypublishing.org
Hodgkins lymphoma, and acute and chronic myeloid leu- of any of several centrosome components including Plk4 results
kaemia [2 7]. Abnormalities have been detected at both in microcephaly [3941]. To study the effects of elevating Plk4
early [810] and advanced stages of disease where they expression in the mouse brain, Marthiens et al. [42] generated
generally indicate poor prognosis [11]. Amplified centrosomes transgenic animals in which Plk4 expression could be activated
also correlate with metastasis of head and neck, prostate and in the central nervous system in response to a tissue-specific
breast tumours [1214]. However, despite these long-known Cre-mediated recombination event. This led to a microce-
associations, the contribution of centrosomal amplification to phaly-like condition that was ascribed to the poor clustering
oncogenesis remains unclear. of amplified centrosomes leading to abnormal mitoses and con-
Centrosome number is normally tightly controlled within sequent apoptosis. Cell death could be overcome by removing
the cell division cycle. At the core of centrosomes lie the nine- p53 function, leading to the accumulation of aneuploidy cells

Open Biol. 5: 150209


fold symmetrical centrioles. Cells enter mitosis with two that would differentiate rather than proliferate.
pairs of orthogonally arranged centrioles that separate to We wished to examine the consequences of elevated Plk4
form the two centrosomes at the poles of the spindle. The expression, and thereby centrosome amplification, in other
centrioles disengage upon the completion of mitosis tissues in the mouse. To have temporal control on the over-
and enable both individual centrioles to initiate the dupli- expression of Plk4, we have developed a mouse line in
cation process. Centriole duplication is controlled by which Plk4 is under the control of a doxycycline-inducible
Polo-like-kinase-4 (Plk4) that phosphorylates the centriole promoter. Induction of Plk4 expression in this mouse leads
protein Stil/Ana2 allowing it to recruit Sas6, the core com- to an early onset of tumour formation in p53 null mice,
ponent of the centriole cartwheel [15 18]. Elevating Plk4 behavioural defects suggesting abnormalities of brain devel-
expression through its ectopic expression or by eliminating opment in agreement with a previous study [39], and
the SCF ubiquitin-protein ligase required for Plk4 destruction hyperproliferation of cells in the pancreas and in the skin.
results in the formation of multiple centrosomes [19 23]. Here we focus upon characterizing defects in the skin of these
The loss of centrioles has different consequences in differ- animals and show that elevated Plk4 leads to amplification of
ent organisms and tissues. Drosophila can tolerate centriole centrosomes and loss of primary cilia. Together this leads to
loss in some, but not all, tissues, allowing defective cell div- both hyperproliferation and uncontrolled differentiation of
isions to continue [23 27]. However, centrioles also serve the basal epidermis. We discuss how these phenotypes can
as basal bodies, the foundations of cilia and flagellae arise and how we might account for the enhancement of these
[28,29], and so are essential to fashion the flys sensory phenotypes when the p53 gene is deleted.
organs for correct physical coordination [24,30]. In mamma-
lian cells, the physical removal of centrosomes prevents cell
cycle progression but eventually centrioles reform by a 2. Results
de novo pathway and the cell cycle resumes [31 33]. In the
mouse, there is a greater reliance on centrioles to generate pri- 2.1. Elevated Plk4 expression dramatically advances the
mary cilia essential for many types of cell signalling.
However, unlike mutants that lack cilia, mutant embryos
onset of tumour formation in p53-deficient mice
deficient for the centriole component Sas4 and thereby lack- Centrosome amplification has been identified as a marker of
ing centrioles exhibit extensive apoptosis associated with poor prognosis of aggressive, drug-resistant tumours in breast,
elevated p53 expression [34]. Apoptosis was rescued in pancreatic and colorectal cancer patients [43,44], but the relation-
embryos double mutant for Sas4 and p53, thus identifying ship of centriole amplification to oncogenesis is uncertain. To
a p53-dependent apoptotic pathway triggered by loss of cen- address this in a model system, we generated a transgenic
trioles. This has been further supported by experiments to mouse that allows the inducible over-expression of wild-type
eliminate Plk4 activity from cultured cells using either an mPlk4, the master regulator of centriole duplication. The
auxin-inducible degradation system or pharmacological inhi- vector comprised the reverse-tetracycline-controlled transactiva-
bition of the enzyme using a small molecule, centrinone tor (rtTA) linked to a tetracycline-responsive element (TRE)
[33,35]. In both these cases, loss of Plk4 activity results in regulating expression of wild-type mPlk4 and was targeted to
loss of centrioles and a p53-dependent arrest of cell cycle the ROSA26 locus. This enabled the inducible and reversible
progression, the mechanism of which is not understood. over-expression of Plk4 at any time of development in the result-
The consequences of Plk4 over-expression also vary in ing transgenic animals or in cultured cells derived from them
different organisms and in different cell types. Over-expression (figure 1a). We designate the homozygous transgenic mouse
or stabilization of Plk4 in either cultured Drosophila cells or harbouring the inducible extra copy of wild-type Plk4 as
mammalian cells leads to multiple centrosomes [19,2123,36] Plk4OE/ Plk4OE.
and in fertilized Drosophila eggs drives the formation of thou- As p53 was reported to overcome both the cell cycle arrest
sands of centrioles at the expense of the normal progression of associated with loss of centrioles resulting from deletion of
nuclear division cycles [20]. Strikingly this also happens in Sas4 or degradation or inhibition of Plk4 [3335] and
unfertilized eggs in which centrioles have been naturally elimi- the cell death resulting from elevated Plk4 expression in the
nated during oogenesis and in which there is no incoming mouse brain [42], we were prompted also to determine the
sperm to provide a basal body. Thus, in this circumstance, cen- consequences of centrosome amplification induced by Plk4
triole formation is entirely driven by Plk4. Moreover, elevated over-expression in a p53 knockout (KO) background (from
expression of Plk4, and indeed perturbation of centrosome now on p53KO). These mice show accelerated tumour for-
function through several routes, can promote tumourigenesis mation, behavioural defects and cell hyperproliferation
in flies [37,38]. associated with elevated Plk4 expression in several tissues
Downloaded from http://rsob.royalsocietypublishing.org/ on May 27, 2017

(a) (b) survival curve (c) malignant lymphomas sarcomas 3


Plk4 transgene 100
100

rsob.royalsocietypublishing.org
Rosa26 Rosa26
locus 5 SA rtTA TRE mPLK4 puro locus 3
Plk4OE /Plk4OE 80

% of tumour free

% of tumours
Plk4OE/Plk4OE (+DOX)
DOX 60
50 Plk4OE/Plk4OE;
p53KO/p53KO 40
Rosa26
locus 5 SA rtTA TRE mPLK4 puro
Rosa26
locus 3 Plk4OE/Plk4OE;
p53KO/p53KO (+DOX) 20
**p < 0.01
+DOX
0 0
0 5 10 15 20 25 30 35 Plk4OE /Plk4OE; Plk4OE /Plk4OE;
time (weeks) p53KO/p53KO p53KO/p53KO (+DOX)

(d ) thymus (e) lymphoma (f) kidney (g) heart

Open Biol. 5: 150209


Plk4OE/Plk4OE: p53KO/p53KO

Li
Ap
Vn *
Li

Li
50 mm 80 mm 120 mm 400 mm

(l) 1 pair of 2 pairs of


(h) +DOX (i) +DOX ( j) +DOX 50 centrioles centrioles
Plk4OE/Plk4OE; p53KO/p53KO

>2 pairs of
centrioles
G 40
M
G MM

% of cells
30
G

20

10
250 mm MM 100 mm 50 mm

DNA g-tubulin DNA Ac-tubulin DNA g-tubulin


(k)
(m) mitotic progression (n) abnormal mitotic cells
Plk4OE /Plk4OE; p53KO/p53KO

35
monopolar
70 prometaphase
30
60 tripolar
prometaphase 25
bipolar 10 mm 10 mm monopolar tripolar 10 mm prometaphase
% of cells

% of cells

50
metaphase 20 anaphase
DNA Ac-tubulin DNA g-tubulin DNA g-tubulin 40 missegregration
anaphase
15
30
20 10

10 5
10 mm 10 mm 10 mm 0 0
chromosome lagging aneuploidy

Figure 1. Tumour formation following tetracycline-inducible conditional Plk4 expression. (a) Doxycycline associates with the rtTA that binds the TRE, leading to transcriptional
activation of Plk4. (b) Tumour incidence in Plk4 transgenic mice in wild-type background (Plk4OE/Plk4OE) or p53 null background (Plk4OE/Plk4OE; p53KO/p53KO) with or without
treatment with doxycycline (DOX) to promote Plk4 over-expression. The differences observed between Plk4OE/Plk4OE; p53KO/p53KO (n 24) and Plk4OE/Plk4OE; p53KO/p53KO
DOX (n 14) survival curves are significant (**p , 0.01; Students t-test). (c) Proportions of sarcomas and lymphomas. Note that animals that developed sarcomas also
showed lymphomas. (d) Architecture of thymus and lymph nodes is obliterated and replaced by sheets of large, round cells with vesicular nuclei (Vn, black arrows). Apoptotic
cells were also present (Ap, black asterisk). (e) Multicentric high-grade large cell lymphoma. (f) Kidney has normal architecture but contains multifocal cortical interstitial and
sub-capsular infiltrates of large lymphocytes (Li). (g) Large sheets of lymphocytes (Li) attached to pericardial surface of heart wall. (hj) These tumours are sarcomas isolated
from Plk4OE/Plk4OE; p53KO/p53KO mice. Typically, large masses of cells extended into muscle fibre bundles (M) and into attached adipose surrounding nerves and blood vessels.
(i,j) Higher magnifications of sarcomas showing pleomorphic and anaplastic cells. Examples of giant (G) or multinucleated (MM) cells are indicated. These tumours were found
close to the front limbs with high percentage of mitotic cells (an average of 510/40 field). (k) Paraffin section of samples from sarcomas where stained to reveal g-tubulin or
acetylated-tubulin (green). DNA is shown in red. Three different samples were analysed and showed a high mitotic index (8.55 + 2.53%, n 1400 cells/sample) in agree-
ment with histological analysis made after H&E staining. (l ) Proportion of cells that show one pair or two pairs of centrioles per cell or show centriole/centrosome amplification
(more than 2 pairs of centrioles). (m) Mitotic progression in sarcomas from cryostat sections. (n) Proportions of mitotic abnormalities in sarcomas. Quantification in
(ln) performed in three different sarcomas; 5001000 cells analysed per sarcoma.

including the pancreas and skin. Here we describe some key parallel studies on the viability of the Plk4OE/Plk4OE line
features of mice that are expressing elevated levels of Plk4 with or without the addition of doxycycline (DOX) to pro-
and focus upon how this affects development of the skin mote Plk4 over-expression. Plk4OE/Plk4OE and Plk4OE/
and pancreas. Plk4OE (DOX) mice remained healthy during the period
We first wished to address the effects of Plk4 over- of study. Litter sizes were reduced in Plk4OE/Plk4OE
expression upon tumour formation and so carried out (DOX), but tumour formation was not observed during
Downloaded from http://rsob.royalsocietypublishing.org/ on May 27, 2017

the first 35 weeks (figure 1b). We found that approxima- centrosome number [33,35,42], we also examined the effects 4
tely one half of Plk4OE/Plk4OE; p53KO/p53KO mice on a of Plk4 over-expression on the pancreas in p53 null mice.

rsob.royalsocietypublishing.org
doxycycline-free diet developed tumours by the age of 20 Plk4 over-expression now resulted in a more than doubling
weeks and all had tumours by 35 weeks. This time course in the diameters of the islets although the cell density was
of tumour appearance parallels previous reports for p53 not as high as when p53 was present (figure 2e,f ). Again,
homozygous null mice [45]. Interestingly, when Plk4 there was an increase in both a- and b-cells in similar pro-
over-expression was induced in the Plk4OE/Plk4OE; p53KO/ portions (89.8 + 3.2% b-cells versus 85.5 + 3.1% in Plk4OE/
p53KO mice by the addition of DOX from eight weeks Plk4OE; p53KO/p53KO without and with DOX). The size of
onwards, tumour formation was accelerated; half of the the islets directly correlated with the levels of Plk4 transcripts
mice developed tumours by 15 weeks and all died within (figure 2g) and immunostaining of the islets revealed elev-
26 weeks (red line; figure 1b). ated Plk4 protein following treatment of the mice with
The tumours arising in Plk4OE/Plk4OE; p53KO/p53KO mice doxycycline (figure 2g,h,j) and an increase in centrosome

Open Biol. 5: 150209


were highly similar whether or not doxycycline was adminis- number (figure 2h,i). Plk4 was present in punctate bodies
tered and comprised mainly lymphomas and sarcomas corresponding to centrosomes, and these increased in the
(figure 1c). The lymphomas invaded a variety of tissues number from one such body in non-doxycycline-treated,
including the thymus, kidney and heart (figure 1dg). How- p53 / cells to four or more in p53 null cells over-
ever, a wide variety of sarcomas arose more frequently and expressing Plk4 (figure 2j ). Thus, elevated expression levels
earlier after Plk4 over-expression. All animals that succumbed of Plk4 and higher centrosome numbers correlate with
to sarcomas also had lymphomas. The sarcomas were typically hyperproliferation of a- and b-cells in the pancreas that is
localized close to the front limbs and developed very rapidly exacerbated in the absence of p53.
within one week. These tumours had highly pleomorphic
and anaplastic cells that displayed a high incidence of mitosis
(an average of 510 mitotic figures per 40 field) suggesting 2.3. Elevated Plk4 over-expression affects melanocyte
they are highly malignant (figure 1hj) and centrosome amplifi-
cation was detected in 34.79 + 5.48% of cells from sarcomas
differentiation
(figure 1l ). The mitotic figures in these Plk4 over-expressing A striking feature of the Plk4OE/Plk4OE mice was skin lesions
tumour cells were typically bipolar with multiple centrosomes that include alopecia at the time of weaning followed by
at the poles (71%). Centrosome clustering was lost in 12.7 + regrowth of hair around one month after birth. Animals
10.7% of prometaphase cells that were multipolar (figure 1kn). that did not lose hair had grey coats (figure 3a,b). Most of
Although we failed to detect any multipolar anaphases, we the grey hair did not persist and there was a regain of
found chromatid lagging in 23.4 + 10.6% of anaphase figures some black fur within the first month. This phenotype led
(figure 1n). It will be of future interest to identify and characterize us to examine the effects of Plk4 over-expression upon
the mesenchymal cells in which these tumours arise. melanocytes and the melanin pigment produced by
We conclude that Plk4 over-expression significantly them. Melanocytes are located in the interfollicular epidermis
advances the onset of tumour formation in p53 null mice and and within hair follicles, and differentiate from neural-
that this is associated with an increased frequency of mitoses crest-derived melanocyte stem cells (MSCs). These MSCs
that generate aneuploid cells characteristic of many tumours. are intermingled with hair follicle stem cells in the bulge
and the hair germ. Differentiated melanocytes produce and
transfer melanin to the adjacent keratinocytes during the
2.2. Elevated Plk4 expression induces hyperproliferation anagen (growth) phase of the cyclical process of hair regener-
ation (reviewed in [46]). Using Fontana-Masson staining, we
of cells in the pancreas found a reduction of melanin granules in the bulge of
The above findings raised the question of whether we could 2-day-old Plk4OE/Plk4OE mice and their apparent absence
identify any tissues exhibiting common cellular changes from follicles, both in a wild-type or p53KO/p53KO back-
that could be associated with events anticipating tumour ground (figure 3d). We also stained skin sections to
development particularly in the absence of p53 function. examine melanin distribution at 20 days. This revealed a
Systematic histological examination of the tissues of reduction in melanin in Plk4OE/Plk4OE mice irrespective of
Plk4OE/Plk4OE mice revealed that although pancreases had whether animals were treated with doxycycline and that
normal lobular architecture with intact acinar cell clusters, was accentuated if the animals were also null for p53
there was enlargement of the islets of Langerhans when (figure 3e). As an alternative way to assess melanin pro-
over-expression of Plk4 was induced (figure 2ac,e). The duction, we determined the relative levels of transcripts for
diameter of the islets was increased by approximately 30% tyrosinase, the enzyme catalysing the first step in melanin
and the density of cells within the islets more than doubled synthesis and which is expressed in melanocytes [47]. Tyrosi-
under these conditions (figure 2ef ). To determine whether nase mRNA was reduced both in untreated Plk4OE/Plk4OE
this reflected differential proliferation of the major two endo- mice that show some elevation of basal Plk4 mRNA levels
crine cell types, we stained pancreas sections with antibodies and following doxycycline treatment to induce Plk4
against glucagon and insulin to detect a- and b-cells, respect- expression. Tyrosinase transcripts were further reduced in a
ively. This revealed a proportionate increase in the number p53 null background (figure 3c). Taken together these obser-
of both glucagon- and insulin-positive cells following induc- vations point to reduced melanin biosynthesis in response to
tion of Plk4 over-expression (82.8 + 6.7% b-cells versus elevated Plk4, an effect enhanced in the absence of p53 so
79.8 + 6.0% in Plk4OE/Plk4OE islets without and with DOX, accounting for the grey hair of the animals.
respectively; figure 2df ). Because loss of p53 is known We then wanted to determine whether the grey hair was a
to exacerbate the effects of both decreased and increased consequence of reduced melanin production or a reduced
Downloaded from http://rsob.royalsocietypublishing.org/ on May 27, 2017

+DOX 5
+/+ Plk4OE/Plk4OE Plk4OE /Plk4OE; p53KO/p53KO
I
(a) (b)

rsob.royalsocietypublishing.org
(c)

AC
AC l

AC
l
l

AC
50 mm AC 50 mm 150 mm

Open Biol. 5: 150209


Plk4OE/Plk4OE Plk4OE /Plk4OE; p53KO/p53KO

(d ) + DOX + DOX
glucagoninsulinDNA

50 mm 50 mm 50 mm 50 mm

(i) Plk4OE/Plk4OE Plk4OE /Plk4OE; p53KO/p53KO


(e) (f)
300 ** +DOX +DOX
DNA E-cadherin

250 160 **
diameter of islets of

no. islet cells 100 mm2


Langerhans (mm)

200
120
Cep 192

** **
150 *
80 10 mm 10 mm 10 mm 10 mm
100
40
50
Cep192

0 0
(g) 14
***
relative Plk4 mRNA levels

12
10 Plk4OE /Plk4OE
in pancreas

8 Plk4OE /Plk4OE +DOX


6 ** Plk4OE /Plk4OE; p53KO/p53KO
4 Plk4OE/Plk4OE; p53KO/p53KO ( j) Plk4OE/Plk4OE Plk4OE /Plk4OE; p53KO/p53KO
2 +DOX
+DOX +DOX
0
(h)
DNA Plk 4

100

80
*
60
% of cells

*** *** ***


40 10 mm 10 mm 10 mm 10 mm

20

0
1 pair of 2 pairs of >2 pairs of
20 dots dots dots

Figure 2. Plk4 over-expression leads to hyperplasia of the pancreatic islets. (ac) Haematoxylineosin-stained sections of mouse pancreas. (a) Control (/) pancreas
shows normal acinar cells (AC), islets of Langerhans (I) and lobular architecture. (b) Pancreas of Plk4OE/Plk4OE (DOX) male has normal lobular architecture, intact acinar
cell clusters within lobes and islets of variable size. Lymphocytes are seen surrounding islets and between clusters of acinar cells (black arrows). (c) Pancreas from a
Plk4OE/Plk4OE; p53KO/p53KO male showing normal lobular architecture and very large islets. (d) Immunofluorescence of pancreas cryosections showing b-cells detected by
anti-insulin (red) and a-cells detected by anti-glucagon (green) in islets. DNA is blue. (e) Diameter of islets (mean + s.d., n 12) in mice of indicated genotypes
without or with (DOX) doxycycline treatment. (f) Density of insulin or glucagon-positive cells (mean + s.d., n 12) in islets of indicated genotypes without and
with doxycline (DOX) treatment. (g) Q-RT-PCR analysis of relative levels of Plk4 transcripts in pancreatic extracts of indicated genotypes without or with (DOX)
doxycycline treatment. Average from three biological samples (three replicates for each). (h) Proportion of cells showing 1 (one pair of dots), 2 (two pairs of dots)
centrosomes or centrosome amplification (more than two pairs of dots) in pancreatic islets. Centrosomes quantified by counting punctate Cep192 or g-tubulin staining.
(i) Pancreas cryosections stained to reveal E-cadherin (green), centrosome component Cep192 (red in merge; white in monochrome) and DNA (blue). Cell borders
identified by E-cadherin outlined in monochrome image. Note: increase in the number of centrosomes/cell following treatment with doxycycline (DOX). ( j) Pancreas
cryosections stained to reveal Plk4 (green) and DNA (blue). Note: increase in the number per cell and size of anti-Plk4-stained dots following treatment with doxycycline
(DOX). Significance was determined by Students t-test. *p , 0.05, **p , 0.01, ***p , 0.005.
Downloaded from http://rsob.royalsocietypublishing.org/ on May 27, 2017

(a) (b) (c) ** ** ***


6
3.0

relative mRNA levels in skin


2.5

rsob.royalsocietypublishing.org
2.0

1.5
tyrosinase
1.0
** *** *** Plk4
0.5

+/+

Plk4OE/PlkOE

Plk4OE/PlkOE
+DOX

Plk4OE/PlkOE;
p53KO/p53KO

Plk4OE/PlkOE;
p53KO/p53KO
+DOX
PlkOE/PlkOE Plk4OE/Plk4OE; p53KO/p53KO
(d) +/+
+DOX +DOX

Open Biol. 5: 150209


2 days

50 m 50 m 50 m 50 m 50 m

Plk4OE/Plk4OE Plk4OE/Plk4OE; p53KO/p53KO


(e) +/+ +DOX +DOX
20 days

50 m 50 m 50 m 50 m 50 m

+/+ Plk4OE/Plk4OE
(f) DNAMITF (g) DNAMITF DNAMITF +DOX
KITDCT MITFKIT KIT DCT KITDCT MITFKIT KIT DCT KITDCT MITFKIT KIT DCT

Bu Bu Bu

10 m 10 m 10 m

Plk4OE/Plk4OE; p53KO/p53KO
(h) DNAMITF DNAMITF +DOX (i)
KITDCT MITFKIT KIT DCT KITDCT MITFKIT KIT DCT
8 +/+
bulb
MIFT+c-KIT+DCT+ cells/

6 Plk4OE/PlkOE
*
** Plk4OE/PlkOE
4 +DOX
Plk4OE/PlkOE;
Bu 2 p53KO/p53KO
Bu
Plk4OE/PlkOE;
0
10 m 10 m p53KO/p53KO
+DOX

Figure 3. Plk4 over-expression leads to loss of hair and its pigmentation. (a) Plk4OE/Plk4OE; p53KO/p53KO mouse exhibiting typical hair loss phenotype. (b) Plk4OE/Plk4OE;
p53KO/p53KO mice showing varying degrees of greying hair. (c) Q-RT-PCR analysis of Plk4 and tyrosinase transcripts in extracts of total back skin from mice of indicated
genotypes and treatment indicating mean + s.e. for three independent biological samples and three replicates/sample (DOX indicates doxycycline treatment).
(d) Fontana-Masson staining of cryosections of P2 back skin from mice of indicated genotypes. DOX indicates treatment with doxycycline. (e) Fontana-Masson staining
of P20 back skin cryosections from mice of indicated genotypes and treated with doxycycline where indicated (DOX). (f) Anagen hair follicle in wild-type back skin
immunostained to reveal melanocyte markers KIT (red in merge; white, monochrome), MITF (green) and DCT (blue in merge; white in monochrome). DNA is represented
in white. Differentiated melanocytes identified by triple KITMITFDCT staining. (g) P2 back skin from Plk4OE/Plk4OE mice without or with Plk4 over-expression
(DOX) showing anagen hair follicle immunostained to reveal melanocyte markers KIT (red in merge; white in monochrome), MITF (green) and DCT (blue in
merge; white in monochrome). DNA is white in merge. Note reduction in differentiated melanocytes identified by triple KITMITFDCT-positive immunostaining.
(h) P2 back skin from Plk4OE/Plk4OE; p53KO/p53KO mice without or with Plk4 over-expression (DOX) showing hair follicles immunostained for melanocyte markers
KIT (red in merge; white in monochrome), MITF (green) and DCT (blue in merge; white in monochrome). DNA is white in merge. Differentiated melanocytes
(KITMITFDCT) are reduced in Plk4OE/Plk4OE; p53KO/p53KO (DOX) mice. (i) Mean numbers + s.d. of differentiated melanocytes (KITMITFDCT) per bulb in
indicated genotypes and treatments. Significance determined by Students t-test. *p , 0.05, **p , 0.01 and ***p , 0.005.
Downloaded from http://rsob.royalsocietypublishing.org/ on May 27, 2017

number of melanocytes undergoing activation/differentiation. positive cells) layers, induction of Plk4 expression in either 7
To this end, we carried out immunofluorescence upon cryosec- a wild-type or p53 null background led to an expansion of
K5-positive cells. This was particularly evident in Plk4OE/

rsob.royalsocietypublishing.org
tions of the back skin of animals 2 days after birth (P2) to
detect the MSC population undergoing differentiation Plk4OE; p53KO/p53KO doxycycline-treated mice at 20 days
(figure 3fi). The tyrosine kinase receptor, c-KIT and dopa- (figure 4d), where both Ki67- and K5-positive cells were
chrome tautomerase (DCT) are hallmarks to identify the MSC significantly expanded to suprabasal layers and K6 staining
population (reviewed in [46]). MSCs co-expressing Kit and extended to most of the epidermis and hair follicles. There
microphthalmia-associated transcription factor (MITF), the was also an increase in the number of cells expressing the
master regulator of melanocyte differentiation (reviewed in early differentiation marker, K10.
[46]), identify the differentiated melanocyte subpopulation. We The above findings were mirrored by the quantitation of
found a reduced number of KITMITFDCT cells in the mRNA levels for these markers. Over-expression of Plk4 in
bulge when Plk4 was over-expressed, particularly in a p53KO/ a p53 null background led to a notable increase in the

Open Biol. 5: 150209


p53KO background (figure 3i). This population is responsible expression of the basal marker K14 and a reduction in
for pigmentation of growing hair. However, the number of mRNA levels for involucrin, filaggrin and loricrin, markers
MSCs (expressing DCT and KIT) in the bulge that will renew of late stages of differentiation figure 4b). We also found a
the melanocyte population in subsequent hair cycles did not reduction of the transcript levels of DNp63, a p53 family
vary significantly between the different genotypes and treat- member that controls expansion of epidermal cells and pro-
ments (an average of 4.355 KITDCT cells per bulge). This motes differentiation [51] (figure 4c). This, together with a
strongly suggests that Plk4 over-expression compromises similar reduction in expression of the Cdk inhibitor p21
melanocyte differentiation and subsequent melanin synthesis. (mp21; figure 4c), is compatible with delayed terminal dif-
ferentiation [52 54]. In agreement, with these results,
percentage of Ki67-positive cells is significantly increased in
2.4. Plk4 over-expression affects cell proliferation Plk4OE/Plk4OE; p53KO/p53KO doxycycline-treated mice at
20 days, suggesting increased proliferation sustains the
in epidermis higher number of suprabasal cells in these samples compared
The above experiments also revealed a thickening of the epi- to control (figure 4e,f ).
dermis and some apparently invaginated hair follicles when Taken together our findings indicate that elevated
Plk4 levels were elevated in Plk4OE/Plk4OE; p53KO/p53KO expression of Plk4 results in hyperproliferation of the epidermis.
mice. Thus, we decided to explore the epidermal phenotype Specifically, we observed an expansion of basal progenitor cells
in greater detail. The epidermis is a stratified structure con- in suprabasal layers and a decreased expression of genes associ-
taining self-renewing stem cells within the basal layer that ated with terminal differentiation. These observations accord
express keratins 5 and 14 (K5, K14) and that through with the thickening of the epidermis and disrupted hair follicle
delamination and/or asymmetrical cell division give rise to morphology as a consequence of Plk4 over-expression.
non-proliferative, spinous and granular layers (expressing
keratins 1 and 10 (K1, K10) and Involucrin) and outer
layers of terminally differentiated stratum corneum cells
2.5. Plk4 over-expression induces centriole over-
(reviewed in [48]). The increased cell density within the duplication and primary cilia disappearance
epidermal basal layer of Plk4 over-expressing samples
(figure 3e) prompted us to ask whether cell proliferation
in epidermis
was affected as a result of increased levels of Plk4. We next considered whether the alterations we had observed
To address whether there was an increased number of in the regulation of cell proliferation and differentiation in the
cycling cells within the basal epidermis, we analysed skin epidermis could reflect changes mediated by elevated Plk4
from 2-day-old pups (P2) and 20 day-old pups (P20) using upon the centrosomes and/or primary cilia, both of which
Ki67 as a marker for proliferating cells (figure 4a c). In rely upon centrioles for their correct formation and function.
wild-type skin (/), Ki67-positive cells are solely found To this end, we stained skin sections to reveal acetylated
in the basal layer, as these are the only cycling cells in unper- a-tubulin, a marker of the primary cilia, and Plk4 itself,
turbed circumstances (figure 4a). We found Ki67-positive which associates with centrioles, either in basal bodies or cen-
cells in the basal layer did not increase significantly following trosomes. In the skin of wild-type mice, the great majority of
doxycycline treatment of Plk4OE/Plk4OE mice (n 300/ cells of the basal epidermis showed bodies of Plk4 staining
sample; 59.5 + 19.4% in Plk4OE/Plk4OE; p53KO/p53KO associated with the base of single primary cilia (figure 5a).
versus 63.9 + 9.4% in control (/)). However, in the skin Over-expression of Plk4 during development led to increased
of Plk4OE/Plk4OE; p53KO/p53KO mice, doxycycline treatment numbers of centrosomes in the basal epidermis and a loss of
also led to the appearance of Ki67-positive cells in the supra- primary cilia (figure 5a,c). Loss of primary cilia and mis-posi-
basal layers (n 425 cells/sample, 7.40 + 3.90%) within 2 tioning of centrosomes was particularly dramatic when Plk4
days after birth (asterisk in figure 4a). We also examined over-expression occurred in a p53 null background (12.2 +
the distribution of keratin 6 (K6), which is normally restricted 4.5% of cells with primary cilia in Plk4OE/Plk4OE; p53KO/
to the hair follicles but is also found in the interfollicular epi- p53KO (DOX) versus 35.3 + 4.9% in Plk4OE/Plk4OE;
dermis in conditions of hyperplasia the epidermis [49,50]. p53KO/p53KO; figure 5a,c). We were still able to observe
Strikingly, we found that K6 was widely expressed in basal some residual primary cilia in hair follicles even though
and suprabasal cells after induction of Plk4 expression in these cells had extra centrioles (figure 5b). However, consist-
either a wild-type or p53 null background (arrowheads in ently, only a single primary cilium was formed (figure 5b),
figure 4a). Whereas in normal skin there is a clear separation and these cilia were longer than those in wild-type cells
between basal (K5-positive cells) and suprabasal (K10- (figure 5d). Thus, the effects of elevated Plk4 may differ in
Downloaded from http://rsob.royalsocietypublishing.org/ on May 27, 2017

(a) +DOX +DOX 8


2 days
Wt Plk4OE/Plk4OE Plk4OE/Plk4OE Plk4OE/Plk4OE; p53KO/p53KO Plk4OE/Plk4OE; p53KO/p53KO

rsob.royalsocietypublishing.org
Epi Epi

g-tubulin
Epi B Epi B
Sp
DNA

Ki67
B B Epi *
Der Der
B
Der Der
Der
HF 30 m 30 m 30 m 30 m 30 m
E-cadherin K6 K5

B
SB B
Der B
B B Der
Der Der
Der

30 m HF 30 m HF 30 m 30 m 30 m
HF HF

Open Biol. 5: 150209


HF

B
E-cadherin K6

Der
B B
Der Der

HF HF HF HF
g-tubulin

Sp
DNA

Sp
K10

Sp Sp Sp
30 m B 30 m 30 m 30 m B 30 m
B B B

(b) 3.5 (c)


1.4
* * Plk4OE/Plk4OE Plk4OE/Plk4OE
3.0 Plk4OE/Plk4OE (+DOX) Plk4OE/Plk4OE (+DOX)
1.2
* Plk4OE/Plk4OE; p53KO/p53KO Plk4OE/Plk4OE; p53KO/p53KO
relative mRNA levels

2.5 Plk4OE/Plk4OE; p53KO/p53KO(+DOX) Plk4OE/Plk4OE; p53KO/p53KO(+DOX)


1.0
relative mRNA levels
2.0 *
0.8

1.5
* 0.6
* *
1.0
* 0.4
*
0.5 **
0.2

0 0
Plk4 K5 K14 K1 K10 Fla Inv Loc mp21 DNP63
basal early late differentiation genes
(d) 20 days
DNA g-tubulin Ki67 E-cadherin K6 K5 E-cadherin K6

control control control control


(e) 2.5 *
Sp Plk4OE/Plk4OE
2.0
basal/suprabasal

B p53KO/p53KO
B
Der 1.5
Der
Plk4OE/Plk4OE; p53KO/p53KO

Plk4OE/Plk4OE
1.0 p53KO/p53KO
+DOX
0.5
HF
HF 30 m 30 m HF HF 30 m HF 30 m
0

+DOX +DOX +DOX +DOX


( f ) 70 Plk4OE/Plk4OE
p53KO/p53KO
% of cells ki67+

Sp
50
B Plk4OE/Plk4OE
B B
Der p53KO/p53KO
Sp Der Der 30
+DOX

B 10 *

30 m HF 30 m 0
30 m 30 m HF HF HF HF HF basal supra
basal

Figure 4. Plk4 over-expression leads to hyperproliferation and abnormal differentiation of cells in suprabasal layers. (a) Immunofluorescence of cryosections of P2
back skin from mice of indicated genotypes treated with doxycycline as indicated (DOX). Note: Ki67 reveals proliferating cells in basal and suprabasal (white
asterisk) epidermal layers of Plk4OE/Plk4OE; p53KO/p53KO (DOX) mice; K6, restricted to hair follicles in controls, is present in epidermis following Plk4 induction
(arrowheads). Basal layer marker K5 present throughout epidermis following Plk4 induction; distribution of early differentiation marker, K10 not affected and does
not reveal differences between the experimental samples and the control at this stage. B, basal; Der, dermis; Epi, epidermis; Sp, suprabasal; dotted lines, dermo-
epidermal border. (b) Quantitative RT-PCR assays for levels of indicated transcripts in total back skin from 2 day pups of indicated genotypes and treatment (n 3
and three replicates for each sample). Note: elevated Plk4 transcripts consistently correlate with low expression of late differentiation genes filaggrin (Fla), involucrin
(Inv) and loricrin (Loc). (c) Quantitative RT-PCR assays as in (b) for P21 (mp21) and DNP63. (d) Immunofluorescence analysis of back skin from 20 day pups of
indicated genotypes and treatment. Note: Ki67-positive cells in basal and suprabasal cells (arrowheads) and K6 in multiple layers co-localizing with K5 (white arrow)
in Plk4OE/Plk4OE; p53KO/p53KO (DOX) mice; expanded staining of K5 and K10. (e) Ratio of basal to suprabasal cells indicated as mean + s.d. ( f ) Proportion of
cycling cells in epidermis. Mean + s.d. % of cells showing Ki67-positive immunostaining. Significance determined by Students t-test. *p , 0.05.
Downloaded from http://rsob.royalsocietypublishing.org/ on May 27, 2017

(a) 9
+DOX

rsob.royalsocietypublishing.org
wild-type (C57BL/6) Plk4OE/Plk4OE Plk4OE/Plk4OE; p53KO/p53KO

DNA Plk4 Act-tubulin


B

EPi
B B
HF
dermis
20 m 20 m 20 m

Open Biol. 5: 150209


dermis
DNA Plk4 Act-tubulin

B
10 m 10 m 10 m
Plk4 Act-tubulin

10 m 10 m 10 m

(b) DNA Plk4 Ac-tubulin Plk4 Ac-tubulin (c) +/+


100 Plk4OE/Plk4OE
Plk4OE/Plk4OE (+DOX)
Plk4OE/Plk4OE; p53KO/p53KO
% cells of epidermis

80 Plk4OE/Plk4OE; p53KO/p53KO (+DOX)


wild-type

***
60 ***
***

40
***
20
+DOX +DOX
0
Plk4OE/Plk4OE

1 pair 2 pairs >2 pairs


of centrioles of centrioles of centrioles
(d)
+/+
Plk4OE/Plk4OE (+DOX)
Plk4OE/Plk4OE; p53KO/p53KO (+DOX)

***
2.0 **
Plk4OE/Plk4OE; p53KO/p53KO

length of cilium (m)

+DOX +DOX
1.5

1.0

0.5

Figure 5. Over-expression of Plk4 leads to multiple centrosomes and loss of primary cilia. (a) Immunostaining of back skin of P2 pups of indicated genotypes and
treatment to reveal Plk4 (red), acetylated tubulin (green) and DNA (blue). Arrows indicate individual primary cilia (anti-acetylated tubulin) that are apically oriented
in basal and suprabasal cells in wild-type control. Note: induction of Plk4 leads to extra centrioles and loss of primary cilia. (b) Immunostaining of hair follicles in
mice of indicated genotypes and treatment. Note: primary cilia (arrowheads) are still observed in hair follicles after Plk4 induction although they appear longer than
in control. (c) Quantification of centriole pairs in basal and superbasal layers. Mean + s.d., n 30 50 cells/condition. (d ) Quantification of primary cilium length
in samples illustrated in (b). Mean + s.d., n 30 50 cells per condition. Significance determined by Students t-test. **p , 0.01, ***p , 0.005.
Downloaded from http://rsob.royalsocietypublishing.org/ on May 27, 2017

+DOX 10
(a) (b)
+/+ Plk4OE/Plk4OE Plk4OE/Plk4OE Plk4OE/Plk4OE; p53KO/p53KO
Wt (+/+)

rsob.royalsocietypublishing.org
2 3
Plk4OE/Plk4OE
centrin2/3 g-tubulin

Plk4OE/Plk4OE +DOX
DNA Plk4

1 1 45 Plk4OE/Plk4OE; p53KO/p53KO +DOX


2 3
**
40
** **
35
2
1 2
10 mm 10 mm 1 10 mm 10 mm

% of keratinocytes
30

25
g -tubulin centrin 2/3 Plk4

g -tubulin centrin 2/3 Plk4

g -tubulin centrin 2/3 Plk4

g -tubulin centrin 2/3 Plk4


20
1 2 1 2 3 1 2 1 2 3
15

Open Biol. 5: 150209


1 2 1 2 3 1 2 1 2 3 10

1 3 mm 2 3 mm 1 3 mm 2 3 mm 3 3 mm 1 3 mm 2 3 mm 1 3 mm 3 mm 2 3 mm 3 0
>4 centrioles

(d)
Wt (+/+)
+ DOX Plk4OE/Plk4OE
45
(c) +/+ Plk4OE/Plk4OE Plk4OE/Plk4OE Plk4OE/Plk4OE; p53KO/p53KO Plk4OE/Plk4OE +DOX
40
35 Plk4OE/Plk4OE; p53KO/p53KO +DOX

% of keratinocytes
30
act-tubulin
DNA Plk4

25
20
** **
15
10

5 mm 5 mm 5 mm 5 mm 5
0
1 cilium >1 cilium

1
(e) Wt (+/+)
4.0
Plk4

Plk4OE/Plk4OE
3.5 **
*

length of cilium (mm)


3.0
2
2.5
2.0
1.5
1.0
Plk4

0.5
1 2
3 mm 3 mm 3 mm 3 mm 0
Plk4OE/Plk4OE +DOX)
Plk4OE/Plk4OE; p53KO/p53KO +DOX

Figure 6. Over-expression of Plk4 leads to multiple centrosomes and loss of primary cilia in primary keratinocytes. (a) Primary culture of keratinocytes isolated from
Wt, Plk4OE/Plk4OE and Plk4OE/Plk4OE; p53KO/p53KO dorsal skin under low calcium conditions. Plk4 over-expression promoted by addition of doxycycline (DOX). Immu-
nostaining reveals Plk4 (red), centrin 2/3 (green) and g-tubulin (blue). DNA stained with DAPI (white). Individual regions magnified in insets showing example of
centriole over-duplication after Plk4 over-expression. (b) Quantitation of centrioles in three independent experiments. Mean values + s.d.; more than 200 cells
quantified for each condition. (c) In vitro, addition of Ca2 induces primary cilia formation in control, Plk4OE/Plk4OE and Plk4OE/Plk4OE; p53KO/p53KO. In control
and Plk4OE/Plk4OE, 38.70 + 3.80% and 31.70 + 2.38% of keratinocytes, respectively, formed a primary cilium after higher Ca2 conditions. If Plk4 is over-
expressed, the percentage of keratinocytes showing primary cilia is reduced to 1/3 in Plk4OE/Plk4OE compared to controls (11.00 + 4.38%). Similar results
were obtained in Plk4OE/Plk4OE; p53KO/p53KO over-expressing Plk4 (9.66 + 4.87%). (d ) Quantitation of percentage of keratinocytes exhibiting one or more primary
cilia under indicated conditions. (e) Quantitation of primary cilium length under indicated conditions. Significance evaluated by Students t-test and compared to
wild-type (*p , 0.05 and **p , 0.01).

different cell types. In the majority of cells of the basal epider- following addition of doxycycline to primary Plk4OE/Plk4OE-
mis, Plk4 over-expression results in elevated numbers of derived keratinocytes cultured in low concentrations of
centrosomes that form at the expense of primary cilia. This calcium reproducibly resulted in centriole over-duplication.
correlates with the increased proliferation of these cells. A pri- (figure 6a,b). By 48 h after addition of calcium, adherens junc-
mary cilium is still able to form alongside additional tions formed and tight junctions and desmosomes were
centrosomes in some hair follicle cells after Plk4 over- assembled. Such cells expressed markers of early and late differ-
expression but these primary cilia are abnormal in structure. entiation and grew on top of each other to form a pseudo-three-
We then prepared primary cultures of keratinocytes from dimensional epidermis. Under these conditions, a single
our mouse lines as these can be cultured under conditions primary cilium formed at the surface of approximately 40% of
that permit either cell proliferation or, following the addi- cells in either wild-type control cells or in Plk4OE/Plk4OE cells
tion of calcium, the formation of cellcell contacts and without Plk4 induction. Addition of doxycycline to the
cellular differentiation. Induction of Plk4 over-expression medium promoted the formation of supernumerary centrioles
Downloaded from http://rsob.royalsocietypublishing.org/ on May 27, 2017

and the formation of fewer primary cilia (figure 6b,c). When chromosome mis-segregation, prolonged time in mitosis 11
a primary cilium was formed, there was only one per cell [33,35], nor the Hippo-signalling pathway, recently shown to

rsob.royalsocietypublishing.org
even though multiple centrioles could be present. Moreover, respond to cytokinesis failure [72], were responsible for trigger-
these primary cilia were significantly longer than in wild-type ing this p53-dependent arrest in a senescent-like G1 state in
cells or Plk4OE/Plk4OE cells that had not been induced to response to centrosome loss. We now show that defects in the
over-express Plk4 (figure 6d). pancreas and skin resulting from Plk4 over-expression are
Thus, over-expression of Plk4 results in supernumerary enhanced by the loss of p53. We do not know the nature of
centrosomes both in cells of the basal epidermis and in cultures this p53-dependent response to either gain or loss of centro-
of primary keratinocytes. In both cases, cells appear to fail to somes but it seems possible that it uses a common pathway as
leave the proliferative state and they fail to form primary both situations can perturb spindle organization and dynamics
cilia. This suggests that over-expression of Plk4 leads to an and cell cycle progression.
alteration in the balance between proliferation and differen- The tumour formation that we observe, however, appears

Open Biol. 5: 150209


tiation of the progenitor epidermal cells. Cilia formation dependent upon the loss of p53 function; it is exacerbated by
could be compromised either directly by the increased Plk4 over-expression but is not seen following Plk4 over-
number of centrosomes or as a consequence of a failure of expression alone. It will be important in future studies to
cells to cease cycling sufficiently to enable ciliogenesis. It is identify the origins of these tumour cells, particularly the sar-
known that the primary cilia receive and transmit extracellular comas that appear with increased frequency when Plk4 levels
signals during development [55 57] and that several ciliary are elevated. That we see tissue hyperplasia rather than neo-
mutants display defects in the commitment of progenitors to plasia in the pancreas and skin might reflect the time course
differentiate. It is also known that lack of primary cilia in of the development of different tumour types in the absence
cells of the basal epidermis would compromise the signalling of p53. Usually, p53 null mice first develop lymphomas and
events required to promote their correct differentiation [58,59], most do not survive for long enough for sarcomas to arise.
so accounting for the defects that we see in cell differentiation Nevertheless, sarcoma formation predominates in mice in
in the skin of Plk4 over-expressing mice. which lymphocytes have been genetically eliminated and it
is conceivable that the development of carcinomas, as
would occur in the skin, would be masked by these earlier
events of lymphoma and then sarcoma formation [73].
3. Discussion The hyperplasia we observe in the pancreas in response to
The great majority of tumour cells are both aneuploid and elevated Plk4 and enhanced in the absence of p53 appears to
have multiple centrosomes [7,8,60], leading to the suggestion affect the differentiated endocrine cells equally. Thus, the
that these properties may be causally related. However, the enlarged islets of Langerhans maintain similar proportions of
extent to which chromosome instability resulting from mul- a- and b-cells suggesting that both populations have prolifer-
tiple centrosomes contributes to tumour formation is not ated and differentiated. In the skin, on the other hand,
clear because centrosome clustering at mitosis usually elevated Plk4 permits proliferating cells to expand into the
ensures the fidelity of chromosome transmission on bipolar suprabasal layers of the epidermis where they show inap-
spindles [61]. The transgenic mouse we have generated propriate expression of differentiation markers. This too is
allows us to induce centriole duplication in response to enhanced in a p53 background. The shift in the balance of
elevated Plk4 and so begin to address the relationship cell proliferation and differentiation could reflect persistence
between multiple centrosomes and tumour formation. Our of the centriole as a structure associated with the centrosome
findings that elevating Plk4 perturbs the balance between and so keeping the cell prepared for cell division. It could
proliferation and differentiation in different tissues in a also result from a loss of barrier function due to the disruption
manner exacerbated by loss of the p53 tumour suppressor in the stratified epidermal architecture as occurs, for example,
gives the potential to identify links between supernumerary in the inflammatory response (reviewed in [74]). The stratifica-
centrosomes and early steps in tumourigenesis. tion of the skin occurs when polarized cells of the basal
Mice deficient for p53 are susceptible to tumour formation epidermis undergo divisions perpendicular to the basal cell
[62] and, moreover, the embryonic fibroblasts established layer to produce the differentiating upper layers [75]. Multiple
from such mice show supernumerary centrosomes [63]. One centrosomes might affect the orientation of mitotic spindles
interpretation of these findings has been that p53 might mediate thus perturbing the mechanism that delivers cells from the
cell cycle arrest in response to increased centrosome numbers as basal to the suprabasal layers in an orderly manner.
part of a tetraploidy checkpoint to monitor completion of cyto- Alternatively, the abnormal differentiation in Plk4 over-
kinesis [64] or by monitoring DNA damage or spindle defects in expressing, p53 null cells could reflect loss of primary cilia
these cells [6567]. Others have proposed a direct role for p53 in that we observe to reciprocate additional centrosome for-
directly regulating centrosome duplication [6870]. While it mation both in cells of the basal epidermis and in cultures
remains difficult to distinguish cause from effect, growing evi- of primary keratinocytes established from this skin. The differ-
dence points towards a p53-mediated response to changes in entiation of cells entering the spinous layer is a Notch
centrosome integrity that receives support from our present signalling-dependent process [76] and a failure of ciliogenesis
study and earlier findings. p53 deletion overcomes either the has been reported to compromise Notch signalling and result
block to cell progression or apoptosis resulting from depletion in defective epidermal differentiation [58]. Thus, in the absence
of several different centrosome proteins in cultured cells [71] of sufficient primary cilia, the signalling pathways required for
or centriole loss in Sas4 knockout mouse embryos [34]. More- correct differentiation would be defective. Reduced signalling
over, recent studies have identified a p53 response that is activity was also previously reported in cultured cells expres-
triggered by the inhibition or the destruction of Plk4 that pre- sing elevated levels of Plk4 [77]. However, in the examples
vents centriole duplication [33,35]. DNA damage, stress, studied by these authors, cells with extra centrosomes often
Downloaded from http://rsob.royalsocietypublishing.org/ on May 27, 2017

Table 1. Primers for mPlk4 and genotyping. 12

rsob.royalsocietypublishing.org
mPlk4RosaFor 50 GGGGACAAGTTTGTACAAAAAAGCAGGCTTCATGGCGGCGTGCATCGGGGAGAGGATCGAGGACTTTAAG30
mPlk4RosaRev 50 GGGGACCACTTTGTACAAGAAAGCTGGGTTTAGGAGTTGGATTAGAAAACATCAGAAGGATGGAAGAAAG30
Plk4Insert_Forward 50 CCGCGCCTGTCCTTTCTCCC30
Plk4Insert_Reverse 50 GTCCGGCCAGGACGACGAGG30
Wt_Rosa_locus 50 GGCAAGCACCACCACTGGCTGGC30
Wt_Rosa_locus 50 GAAGTGTAACTGTGGACAGAGGAGCC30
IMR8306_p53Mut_FW 50 CTATCAGGACATAGCGTTGG30
IMR7778_p53_Rev 50 ATACTCAGAGCCGGCCT30

Open Biol. 5: 150209


IMR7777_p53_FW 50 ACAGCGTGGTGGTACCTTAT30

formed more than one primary cilium. These cilia were associ- elements attR1/attR2. mPlk4 cDNA flanked by attL1/attL2
ated with reduced concentrations of signalling molecules and sites was introduced into the vector by Gateway cloning
this appeared to be responsible for the reduction in signalling (Life Technology), yielding a Rosa26 targeting vector
activity. This contrasts to our findings in keratinocytes where harbouring tetracycline-inducible cDNA for Plk4. Primers
supernumerary centrosomes appear to form at the expense used for subcloning the mousePlk4cDNA were mPlk4Rosa-
of the formation of cilia. The occasional cell that we do find For and mPlk4RosaRev (table 1). The targeting vector was
with two primary cilia of increased length seems at odds introduced into JM8 mESCs by electroporation, and success-
with these earlier findings. The net outcome of disrupted sig- ful integrations were selected for with 1 mg ml21 puromycin.
nalling is the same although in one case it appears to be the Correct targeting was confirmed by long-range PCR across
consequence of the inefficient operation of the signalling the 50 homology arm of clonal puromycin-resistant cell lines.
machinery and in the other through the loss of the mechanical
apparatus, the cilia themselves.
In addition to the work we now report, another study has
4.2. Histological analysis of skin and pancreas
described a p53-dependent response to mitotic abnormalities Samples were fixed in 4% paraformaldehyde in PBS at 48C
resulting from Plk4 over-expression in the developing mouse overnight and then dehydrated and embedded in paraffin
brain [42]. These authors described how this led neural stem wax. Eight micrometre sections were stained with haema-
cells to undertake multipolar mitoses leading to aneuploid toxylin and eosin for histological analysis. Samples from
cells that were directed to p53-dependent apoptosis. They dorsal skin or pancreas were washed in PBS, embedded in
reported that loss of p53 allowed aneuploid cells to accumulate OCT compound and kept at 2808C prior to cryosectioning
and differentiate thereby reducing the proportion of proliferat- for immunofluorescence.
ing cells and reducing brain size. These consequences seem to
differ from the ones we now report in the pancreas and
skin where p53 enhances the effects of Plk4 over-expression 4.3. Fontana-Masson staining
by allowing cells to proliferate. This suggests that the Fontana-Masson staining of backskin cryosections was per-
responses to the supernumerary centrosomes resulting from formed following the protocol provided in the Fontana-Masson
over-expression of Plk4 might differ in different tissues. kit (Abcam, ab150669).
Our study highlights the importance of the p53 pathway
in monitoring defects elicited by the elevated expression of
Plk4. The increase in centrosome numbers and decrease in 4.4. Culture of kerotinocytes
primary cilia strongly suggest that the effects we observe Mouse keratinocytes were isolated from the backskin of 2-
reflect the principal function of Plk4 to drive centrosome day-old pups from each genetic background using dispase
duplication, although we cannot at this stage exclude an and trypsin. After filtration in 40 mm cell strainers, cells
involvement of Plk4 in regulating other aspects of cellular were cultured in low calcium medium (50 mM Ca2) on
physiology (e.g. [78,79]). Further studies are now required plates coated with coating matrix (Gibco) as previously
to dissect out the precise manner by which the p53 pathway described [81]. Once confluency was reached, coverslips
is triggered together with the mechanisms whereby elevated were either fixed in 2208C methanol to visualize centrosomes
Plk4 affects cell cycle progression and cellular architecture, or cultures were shifted to 2 mM Ca2 media for 48 h to
how these might affect the differentiation programme of the analyse cytoskeleton and primary cilium formation.
cell and how this could contribute to tumour development.

4.5. Antibodies
4. Material and methods Primary antibodies were obtained from the following sources:
rabbit anti-Cep192 [82] (1 : 1000), rabbit anti-keratin 5 ab52635
4.1. Generation of mESCs inducibly expressing Plk4 (Abcam, 1 : 1000), chicken anti-keratin 5 #905901 (BioLegend,
1 : 1000), rabbit anti-keratin 10 ab76318 (Abcam, 1 : 1000),
from the Rosa26 locus rabbit anti-keratin 6 #905701 (BioLegend, 1 : 1000), rabbit
The rtTA gene and response element (ClonTech) were cloned anti-cKit ab5506 (Abcam), rabbit anti-Ki67 ab15580
into a Rosa26 targeting vector [80] upstream of Gateway (Abcam, 1 : 500), mouse anti-g-tubulin monoclonal GTU-88
Downloaded from http://rsob.royalsocietypublishing.org/ on May 27, 2017

(Sigma, 1 : 200), rabbit anti-g-tubulin T3559 (Sigma, 1 : 500), rat Table 2. Primers for RT-QPCR. 13
anti-PLK4 [78] (1 : 1000), mouse anti-centrin2/3 S3332 (Santa

rsob.royalsocietypublishing.org
Cruz Biotechnology, 1 : 500), guinea-pig anti-insulin ab7842 Plk4 forward 50 AGGAGAAACTAATGAGCACCACA30
(Abcam, 1 : 50), mouse monoclonal anti-glucagon ab10988
Plk4 reverse 50 TGGCTCTCGTGTCAGTCCAA30
(Abcam, 1 : 200), mouse monoclonal anti-acetylated-tubulin
clone 6-11B-1 (Sigma, 1 : 200), mouse anti-MITF ab80651 GAPDH forward 50 AAGGTCATCCCAGAGCTGAA30
(Abcam, 1 : 200), rabbit anti-DCT ab74073 (Abcam, 1 : 200), GAPDH reverse 50 CTGCTTCACCACCTTCTTGA30
mouse anti-E-cadherin clone ECDD-2 (Invitrogen, 1 : 200), rat K1 forward 50 GAACACTAAGCTGGCCCTGGACAT30
anti-mouse-cKIT clone 2B8 (Biolegend). Secondary antibodies
K1 reverse 50 CCTCGGGAGTAACTGGTGGAAACA30
used (1 : 2000 for immunofluorescence) were conjugated with
Alexa 488, Alexa 568 or Alexa 647 (Invitrogen) and had minimal K5 forward 50 CAGTGTGCCAACCTCCAGAACG30
cross-reactivity to other species. K5 reverse 50 AGCCCGCTACCCAAACCAAGAC30

Open Biol. 5: 150209


K10 forward 50 GGAGGGTAAAATCAAGGAGTGGTA30
4.6. Fixation protocol for tissues and cells K10 reverse 50 TCAATCTGCAGCAGCACGTT30
For identification of centrosomal proteins by immuno- K14 forward 50 GACGCCGCCCCTGGTGTG30
fluorescence, preparations were fixed (12 min in ice-cold K14 reverse 50 GGTGGCGATCTCCTGCTC30
methanol), quickly rinsed in 1PBS and then permeabilized lagrin forward 50 GGAGGCATGGTGGAACTGA30
in 1PBS; 0.1% Triton-X100 for three times, for 5 min each.
lagrin reverse 50 TGTTTATCTTTTCCCTCACTTCTACATC30
Preparations were blocked with 1PBS; 0.1% Tween 20;
10% FBS for 1 h, followed by primary antibody incubation involucrin forward 50 GTCCGGTTCTCCAATTCGTGTTT30
in the same solution for at least 2 h at room temperature. involucrin reverse 50 GCAATTGGAAGAGAAGCAGCATCAG30
Washes were performed in 1PBS; 0.1% Tween 20 for loricrin forward 50 TCACTCATCTTCCCTGGTGCTT30
30 minutes before the addition of secondary antibody in
loricrin reverse 50 GTCTTTCCACAACCCACAGGA30
1PBS; 0.1% Tween 20 with 10% FBS. Preparations were
washed as previously, mounted in Vectashield with DAPI DNp63 forward 50 CTGGAAAACAATGCCCAGAC30
and sealed. DNp63 reverse 50 GAGGAGCCGTTCTGAATCTG30
mp21_forward 50 GTGGGTCTGACTCCAGCCC30
4.7. Microscopy mp21_reverse 50 CCTTCTCGTGAGACGCTTAC30
Images were collected on a Zeiss LSM 510 Meta Laser Scan- tyrosinase forward 50 GCGAAGGCACCGCCCTCTTT30
ning Confocal Microscope using 63X/1.4 or 100X/1.4 oil tyrosinase reverse 50 TCCCACCAGTGCTGCCCCAA30
objectives, and the LSM 510 v. 4.2 software. Images were
deconvolved using HUYGENS PROFESSIONAL software; proces-
sing and analysis was performed with IMAGEJ v. 1.50b and for at least three biological samples and fold changes calcu-
Adobe PHOTOSHOP CS5. lated using the 22DDCT method.

4.8. RT-qPCR conditions


Competing interests. We declare we have no competing interests.
RNA was isolated from the different tissues with RNAqueous
Funding. P.A.C. is supported by a Program Grant from Cancer
Kit (Ambion) and RT-QPCR was performed using Power Research UK to D.M.G. L.B. is the recipient of a Cancer Research
SYBRw Green RNA-to-CTTM 1-Step Kit (Life Technologies) UK research studentship from Cambridge Cancer Centre. M.S. is
on a Stepone Plus 96 RT system (Life Technologies) with supported by an EMBO Fellowship. M.H. is a Wellcome Trust Sir
GAPDH as reference gene. Primers pairs (forward and Henry Dale Fellow, jointly funded by the Wellcome Trust and the
reverse) used for each gene are indicated in table 2. Specificity Royal Society and receiving core support grant from the Wellcome
Trust and Cancer Research UK to the Wellcome TrustCancer
was confirmed by subsequent melting curve analysis or gel Research UK Gurdon Institute. W.C.S. is supported by core funding
electrophoresis. Levels of PCR product were expressed as a to the Wellcome Trust Sanger Institute; M.Z.G. holds a Wellcome
function of GAPDH. Reactions were performed in triplicate Trust Senior Fellowship.

References
1. Scheer U. 2014 Historical roots of centrosome polarity. Proc. Natl Acad. Sci. USA 95, 29502955. 6. Sato N, Mizumoto K, Nakamura M, Nakamura K,
research: discovery of Boveris microscope slides in (doi:10.1073/pnas.95.6.2950) Kusumoto M, Niiyama H, Ogawa T, Tanaka M. 1999
Wurzburg. Phil. Trans. R. Soc. B 369, 20130469. 4. Pihan GA, Purohit A, Wallace J, Knecht H, Woda B, Centrosome abnormalities in pancreatic ductal
(doi:10.1098/rstb.2013.0469) Quesenberry P, Doxsey SJ. 1998 Centrosome defects carcinoma. Clin. Cancer Res. 5, 963 970.
2. Kramer A, Neben K, Ho AD. 2005 Centrosome and genetic instability in malignant tumors. Cancer 7. Giehl M, Fabarius A, Frank O, Hochhaus A, Hafner
aberrations in hematological malignancies. Cell Biol. Res. 58, 39743985. M, Hehlmann R, Seifarth W. 2005 Centrosome
Int. 29, 375383. (doi:10.1016/j.cellbi.2005.03.004) 5. Hsu L-C, Kapali M, DeLoia JA, Gallion HH. 2005 aberrations in chronic myeloid leukemia correlate
3. Lingle WL, Lutz WH, Ingle JN, Maihle NJ, Salisbury Centrosome abnormalities in ovarian cancer. with stage of disease and chromosomal instability.
JL. 1998 Centrosome hypertrophy in human breast Int. J. Cancer 113, 746751. (doi:10.1002/ijc. Leukemia 19, 1192 1197. (doi:10.1038/sj.leu.
tumors: Implications for genomic stability and cell 20633) 2403779)
Downloaded from http://rsob.royalsocietypublishing.org/ on May 27, 2017

8. Lingle WL, Barrett SL, Negron VC, DAssoro AB, Plk4/Sak levels to block centriole reduplication. instability of Polo-like kinase 4 limits centrosome 14
Boeneman K, Liu W, Whitehead CM, Reynolds C, J. Cell Biol. 184, 225 239. (doi:10.1083/jcb. duplication to once per cell cycle. Genes Dev. 26,

rsob.royalsocietypublishing.org
Salisbury JL. 2002 Centrosome amplification drives 200808049) 2684 2689. (doi:10.1101/gad.207027.112)
chromosomal instability in breast tumor 22. Habedanck R, Stierhof Y-D, Wilkinson CJ, Nigg EA. 37. Castellanos E, Dominguez P, Gonzalez C. 2008
development. Proc. Natl Acad. Sci. USA 99, 2005 The Polo kinase Plk4 functions in centriole Centrosome dysfunction in Drosophila neural stem
19781983. (doi:10.1073/pnas.032479999) duplication. Nat. Cell Biol. 7, 1140 1146. (doi:10. cells causes tumors that are not due to genome
9. Pihan GA, Wallace J, Zhou Y, Doxsey SJ. 2003 1038/ncb1320) instability. Curr. Biol. 18, 12091214. (doi:10.1016/
Centrosome abnormalities and chromosome 23. Bettencourt-Dias M et al. 2005 SAK/PLK4 is required for j.cub.2008.07.029)
instability occur together in pre-invasive carcinomas. centriole duplication and flagella development. Curr. 38. Basto R, Brunk K, Vinadogrova T, Peel N, Franz A,
Cancer Res. 63, 1398 1404. Biol. 15, 21992207. (doi:10.1016/j.cub.2005.11.042) Khodjakov AL, Raff JW. 2008 Centrosome
10. Segat D et al. 2010 Pericentriolar material analyses 24. Basto R, Lau J, Vinogradova T, Gardiol A, Woods CG, amplification can initiate tumorigenesis in flies. Cell
in normal esophageal mucosa, Barretts metaplasia Khodjakov AL, Raff JW. 2006 Flies without 133, 1032 1042. (doi:10.1016/j.cell.2008.05.039)

Open Biol. 5: 150209


and adenocarcinoma. Histol. Histopathol. 25, centrioles. Cell 125, 1375 1386. (doi:10.1016/j.cell. 39. Martin C-A et al. 2014 Mutations in PLK4,
551560. 2006.05.025) encoding a master regulator of centriole
11. Yamamoto Y, Matsuyama H, Furuya T, Oga A, 25. Rodrigues-Martins A, Bettencourt-Dias M, Riparbelli biogenesis, cause microcephaly, growth failure and
Yoshihiro S, Okuda M, Kawauchi S, Sasaki K, Naito M, Ferreira C, Ferreira I, Callaini G, Glover DM. 2007 retinopathy. Nat. Genet. 46, 1283 1292. (doi:10.
K. 2004 Centrosome hyperamplification predicts DSAS-6 organizes a tube-like centriole precursor, 1038/ng.3122)
progression and tumor recurrence in bladder cancer. and its absence suggests modularity in centriole 40. Godinho SA, Pellman D. 2014 Causes and
Clin. Cancer Res. 10, 6449 6455. (doi:10.1158/ assembly. Curr. Biol. 17, 1465 1472. (doi:10.1016/ consequences of centrosome abnormalities in
1078-0432.CCR-04-0773) j.cub.2007.07.034) cancer. Phil. Trans. R. Soc. B 369, 20130467.
12. DAssoro AB, Lingle WL, Salisbury JL. 2002 26. Varmark H, Llamazares S, Rebollo E, Lange B, Reina (doi:10.1098/rstb.2013.0467)
Centrosome amplification and the development of J, Schwarz H, Gonzalez C. 2007 Asterless is a 41. Mahmood S, Ahmad W, Hassan MJ. 2011
cancer. Oncogene 21, 6146 6153. (doi:10.1038/sj. centriolar protein required for centrosome function Autosomal recessive primary microcephaly (MCPH):
onc.1205772) and embryo development in Drosophila. Curr. Biol. clinical manifestations, genetic heterogeneity and
13. Pihan GA, Purohit A, Wallace J, Malhotra R, Liotta L, 17, 17351745. (doi:10.1016/j.cub.2007.09.031) mutation continuum. Orphanet. J. Rare Dis. 6, 39.
Doxsey SJ. 2001 Centrosome defects can account for 27. Conduit PT, Wainman A, Raff JW. 2015 Centrosome (doi:10.1186/1750-1172-6-39)
cellular and genetic changes that characterize function and assembly in animal cells. Nat. Rev. 42. Marthiens V, Rujano MA, Pennetier C, Tessier S,
prostate cancer progression. Cancer Res. 61, Mol. Cell Biol. 16, 611624. (doi:10.1038/nrm4062) Paul-Gilloteaux P, Basto R. 2013 Centrosome
2212 2219. 28. Fuller MT. 1993 The development of Drosophila amplification causes microcephaly. Nat. Cell Biol. 15,
14. Reiter R et al. 2009 Centrosome abnormalities in melanogaster. I. Cold Spring Harbor, NY: Cold Spring 731740. (doi:10.1038/ncb2746)
head and neck squamous cell carcinoma (HNSCC). Harbor Laboratory Press. 43. Glinsky GV. 2006 Genomic models of metastatic
Acta Otolaryngol. 129, 205 213. (doi:10.1080/ 29. Chevalier RL. 1969 The fine structure of cancer: functional analysis of death-from-cancer
00016480802165767) campaniform sensilla on the halteres of Drosophila signature genes reveals aneuploid, anoikis-resistant,
15. Ohta M, Ashikawa T, Nozaki Y, Kozuka-Hata H, Goto melanogaster. J. Morphol. 128, 443 463. (doi:10. metastasis-enabling phenotype with altered cell
H, Inagaki M, Oyama M, Kitagawa D. 2014 Direct 1002/jmor.1051280405) cycle control and activated Polycomb group (PcG)
interaction of Plk4 with STIL ensures formation of a 30. Baker JD, Adhikarakunnathu S, Kernan MJ. 2004 protein chromatin silencing pathway. Cell Cycle 5,
single procentriole per parental centriole. Nat. Mechanosensory-defective, male-sterile unc mutants 1208 1216. (doi:10.4161/cc.5.11.2796)
Commun. 5, 5267. (doi:10.1038/ncomms6267) identify a novel basal body protein required for 44. Finetti P et al. 2008 Sixteen-kinase gene expression
16. Dzhindzhev NS, Tzolovsky G, Lipinszki Z, Schneider ciliogenesis in Drosophila. Development 131, 3411 identifies luminal breast cancers with poor
S, Lattao R, Fu J, Debski J, Dadlez M, Glover DM. 3422. (doi:10.1242/dev.01229) prognosis. Cancer Res. 68, 767 776. (doi:10.1158/
2014 Plk4 phosphorylates Ana2 to trigger Sas6 31. Khodjakov AL, Rieder CL. 2001 Centrosomes 0008-5472.CAN-07-5516)
recruitment and procentriole formation. Curr. Biol. enhance the fidelity of cytokinesis in vertebrates 45. Harvey M, McArthur MJ, Montgomery CA, Butel JS,
24, 2526 2532. (doi:10.1016/j.cub.2014.08.061) and are required for cell cycle progression. J. Cell Bradley A, Donehower LA. 1993 Spontaneous and
17. Kratz A-S, Barenz F, Richter KT, Hoffmann I. 2015 Biol. 153, 237 242. (doi:10.1083/jcb.153.1.237) carcinogen-induced tumorigenesis in p53-deficient
Plk4-dependent phosphorylation of STIL is required 32. Mahoney NM, Goshima G, Douglass AD, Vale RD. mice. Nat. Genet. 5, 225 229. (doi:10.1038/
for centriole duplication. Biol. Open 4, 370377. 2006 Making microtubules and mitotic spindles in ng1193-225)
(doi:10.1242/bio.201411023) cells without functional centrosomes. Curr. Biol. 16, 46. Mort RL, Jackson IJ, Patton EE. 2015 The
18. Moyer TC, Clutario KM, Lambrus BG, Daggubati V, 564 569. (doi:10.1016/j.cub.2006.01.053) melanocyte lineage in development and disease.
Holland AJ. 2015 Binding of STIL to Plk4 activates 33. Wong YL et al. 2015 Cell biology. Reversible Development 142, 620 632. (doi:10.1242/dev.
kinase activity to promote centriole assembly. J. Cell centriole depletion with an inhibitor of Polo-like 106567)
Biol. 209, 863878. (doi:10.1083/jcb.201502088) kinase 4. Science 348, 1155 1160. (doi:10.1126/ 47. Wasmeier C, Hume AN, Bolasco G, Seabra MC.
19. Cunha-Ferreira I, Rodrigues-Martins A, Bento I, science.aaa5111) 2008 Melanosomes at a glance. J. Cell. Sci. 121,
Riparbelli M, Zhang W, Laue E, Callaini G, Glover 34. Bazzi H, Anderson KV. 2014 Acentriolar mitosis 3995 3999. (doi:10.1242/jcs.040667)
DM, Bettencourt-Dias M. 2009 The SCF/Slimb activates a p53-dependent apoptosis pathway in 48. Hsu Y-C, Li L, Fuchs E. 2014 Emerging interactions
ubiquitin ligase limits centrosome amplification the mouse embryo. Proc. Natl Acad. Sci. USA 111, between skin stem cells and their niches. Nat. Med.
through degradation of SAK/PLK4. Curr. Biol. 19, E1491 E1500. (doi:10.1073/pnas.1400568111) 20, 847 856. (doi:10.1038/nm.3643)
43 49. (doi:10.1016/j.cub.2008.11.037) 35. Lambrus BG, Uetake Y, Clutario KM, Daggubati V, 49. Weiss RA, Eichner R, Sun TT. 1984 Monoclonal
20. Rodrigues-Martins A, Riparbelli M, Callaini G, Glover Snyder M, Sluder G, Holland AJ. 2015 p53 protects antibody analysis of keratin expression in epidermal
DM, Bettencourt-Dias M. 2007 Revisiting the role of against genome instability following centriole diseases: a 48- and 56-kdalton keratin as molecular
the mother centriole in centriole biogenesis. Science duplication failure. J. Cell Biol. 210, 63 77. (doi:10. markers for hyperproliferative keratinocytes. J. Cell
316, 1046 1050. (doi:10.1126/science.1142950) 1083/jcb.201502089) Biol. 98, 1397 1406. (doi:10.1083/jcb.98.4.1397)
21. Rogers GC, Rusan NM, Roberts DM, Peifer M, Rogers 36. Holland AJ, Fachinetti D, Zhu Q, Bauer M, Verma 50. Stoler A, Kopan R, Duvic M, Euchs E. 1988 Use of
SL. 2009 The SCF Slimb ubiquitin ligase regulates IM, Nigg EA, Cleveland DW. 2012 The autoregulated monospecific antisera and cRNA probes to localize
Downloaded from http://rsob.royalsocietypublishing.org/ on May 27, 2017

the major changes in keratin expression during prevented by centrosomal clustering. Science 307, 72. Ganem NJ, Cornils H, Chiu S-Y, ORourke KP, Arnaud 15
normal and abnormal epidermal differentiation. 127 129. (doi:10.1126/science.1104905) J, Yimlamai D, Thery M, Camargo FD, Pellman D.

rsob.royalsocietypublishing.org
J. Cell Biol. 107, 427 446. (doi:10.1083/jcb. 62. Donehower LA, Harvey M, Slagle BL, McArthur MJ, 2014 Cytokinesis failure triggers hippo tumor
107.2.427) Montgomery CA, Butel JS, Bradley A. 1992 Mice suppressor pathway activation. Cell 158, 833 848.
51. Danilova N, Sakamoto KM, Lin S. 2008 p53 family in deficient for p53 are developmentally normal but (doi:10.1016/j.cell.2014.06.029)
development. Mech. Dev. 125, 919931. (doi:10. susceptible to spontaneous tumours. Nature 356, 73. Landuzzi L et al. 2014 Genetic prevention of
1016/j.mod.2008.09.003) 215 221. (doi:10.1038/356215a0) lymphoma in p53 knockout mice allows the early
52. Deng C, Zhang P, Wade Harper J, Elledge SJ, Leder 63. Fukasawa K, Choi T, Kuriyama R, Rulong S, Vande development of p53-related sarcomas. Oncotarget 5,
P. 1995 Mice Lacking p21CIP1/WAF1 undergo Woude GF. 1996 Abnormal centrosome 11 924 11 938. (doi:10.18632/oncotarget.2650)
normal development, but are defective in G1 amplification in the absence of p53. Science 271, 74. Simpson CL, Patel DM, Green KJ. 2011
checkpoint control. Cell 82, 675684. (doi:10.1016/ 1744 1747. (doi:10.1126/science.271.5256.1744) Deconstructing the skin: cytoarchitectural
0092-8674(95)90039-X) 64. Andreassen PR, Lohez OD, Lacroix FB, Margolis RL. determinants of epidermal morphogenesis. Nat.

Open Biol. 5: 150209


53. Brugarolas J, Chandrasekaran C, Gordon JI, Beach D, 2001 Tetraploid state induces p53-dependent arrest Rev. Mol. Cell Biol. 12, 565 580. (doi:10.1038/
Jacks T, Hannon GJ. 1995 Radiation-induced cell of nontransformed mammalian cells in G1. Mol. nrm3175)
cycle arrest compromised by p21 deficiency. Nature Biol. Cell 12, 13151328. (doi:10.1091/mbc.12.5. 75. Williams SE, Ratliff LA, Postiglione MP, Knoblich JA,
377, 552557. (doi:10.1038/377552a0) 1315) Fuchs E. 2014 Par3-mInsc and Gai3 cooperate to
54. Martn-Caballero J, Flores JM, Garca-Palencia P, 65. Uetake Y, Sluder G. 2004 Cell cycle progression after promote oriented epidermal cell divisions through
Serrano M. 2001 Tumor susceptibility of cleavage failure: mammalian somatic cells do not LGN. Nat. Cell Biol. 16, 758769. (doi:10.1038/
p21(Waf1/Cip1)-deficient mice. Cancer Res. 61, possess a tetraploidy checkpoint. J. Cell Biol. 165, ncb3001)
6234 6238. 609 615. (doi:10.1083/jcb.200403014) 76. Estrach S, Cordes R, Hozumi K, Gossler A, Watt FM.
55. Bershteyn M, Atwood SX, Woo W-M, Li M, Oro AE. 66. Wong C, Stearns T. 2005 Mammalian cells lack 2008 Role of the Notch ligand Delta1 in embryonic
2010 MIM and cortactin antagonism regulates checkpoints for tetraploidy, aberrant centrosome and adult mouse epidermis. J. Invest. Dermatol.
ciliogenesis and hedgehog signaling. Dev. Cell 19, number, and cytokinesis failure. BMC Cell Biol. 6, 6. 128, 825832. (doi:10.1038/sj.jid.5701113)
270283. (doi:10.1016/j.devcel.2010.07.009) (doi:10.1186/1471-2121-6-6) 77. Mahjoub MR, Stearns T. 2012 Supernumerary
56. Goetz SC, Anderson KV. 2010 The primary cilium: a 67. Aylon Y, Michael D, Shmueli A, Yabuta N, Nojima H, centrosomes nucleate extra cilia and compromise
signalling centre during vertebrate development. Oren M. 2006 A positive feedback loop between the primary cilium signaling. Curr. Biol. 22,
Nat. Rev. Genet. 11, 331344. (doi:10.1038/ p53 and Lats2 tumor suppressors prevents 1628 1634. (doi:10.1016/j.cub.2012.06.057)
nrg2774) tetraploidization. Genes Dev. 20, 26872700. 78. Coelho PA, Bury L, Sharif B, Riparbelli MG, Fu J,
57. Santos N, Reiter JF. 2010 Tilting at nodal windmills: (doi:10.1101/gad.1447006) Callaini G, Glover DM, Zernicka-Goetz M. 2013
planar cell polarity positions cilia to tell left from 68. Tarapore P, Horn HF, Tokuyama Y, Fukasawa K. 2001 Spindle formation in the mouse embryo requires
right. Dev. Cell 19, 56. (doi:10.1016/j.devcel.2010. Direct regulation of the centrosome duplication Plk4 in the absence of centrioles. Dev. Cell 27,
07.001) cycle by the p53-p21Waf1/Cip1 pathway. Oncogene 586597. (doi:10.1016/j.devcel.2013.09.029)
58. Ezratty EJ, Stokes N, Chai S, Shah AS, Williams SE, 20, 31733184. (doi:10.1038/sj.onc.1204424) 79. Martindill DMJ, Risebro CA, Smart N, Franco-Viseras
Fuchs E. 2011 A role for the primary cilium in Notch 69. Tarapore P, Tokuyama Y, Horn HF, Fukasawa K. 2001 MDM, Rosario CO, Swallow CJ, Dennis JW, Riley PR.
signaling and epidermal differentiation during skin Difference in the centrosome duplication regulatory 2007 Nucleolar release of Hand1 acts as a molecular
development. Cell 145, 1129 1141. (doi:10.1016/j. activity among p53 hot spot mutants: potential switch to determine cell fate. Nat. Cell Biol. 9,
cell.2011.05.030) role of Ser 315 phosphorylation-dependent 1131 1141. (doi:10.1038/ncb1633)
59. Croyle MJ et al. 2011 Role of epidermal primary centrosome binding of p53. Oncogene 20, 80. Vooijs M, Jonkers J, Berns A. 2001 A highly efficient
cilia in the homeostasis of skin and hair follicles. 6851 6863. (doi:10.1038/sj.onc.1204848) ligand-regulated Cre recombinase mouse line shows
Development 138, 1675 1685. (doi:10.1242/dev. 70. Shinmura K, Bennett RA, Tarapore P, Fukasawa K. that LoxP recombination is position dependent.
060210) 2007 Direct evidence for the role of centrosomally EMBO Rep. 2, 292297. (doi:10.1093/embo-
60. Nam H-J, Chae S, Jang S-H, Cho H, Lee J-H. 2010 localized p53 in the regulation of centrosome reports/kve064)
The PI3 K-Akt mediates oncogenic Met-induced duplication. Oncogene 26, 2939 2944. (doi:10. 81. Nowak JA, Fuchs E. 2009 Isolation and culture of
centrosome amplification and chromosome 1038/sj.onc.1210085) epithelial stem cells. Methods Mol. Biol. 482,
instability. Carcinogenesis 31, 15311540. (doi:10. 71. Mikule K, Delaval B, Kaldis P, Jurcyzk A, Hergert P, 215232. (doi:10.1007/978-1-59745-060-7_14)
1093/carcin/bgq133) Doxsey S. 2007 Loss of centrosome integrity induces 82. Zhu F et al. 2008 The mammalian SPD-2 ortholog
61. Quintyne NJ, Reing JE, Hoffelder DR, Gollin SM, p38-p53-p21-dependent G1-S arrest. Nat. Cell Biol. Cep192 regulates centrosome biogenesis. Curr. Biol.
Saunders WS. 2005 Spindle multipolarity is 9, 160 170. (doi:10.1038/ncb1529) 18, 136 141. (doi:10.1016/j.cub.2007.12.055)

You might also like